A Clinical Quality Management System (CQMS) is a specialized QMS used in clinical research, designed to uphold scientific integrity, regulatory compliance, and participant safety throughout the lifecycle of clinical trials.
Clinical QMS is predominantly used in life science industries, including pharmaceutical, biotechnology, and medical device companies, healthcare institutions, and Contract Research Organizations (CROs).
Clinical QMS is governed by a comprehensive set of international and regional requirements, including ICH E6 (R2) Good Clinical Practice (GCP), ISO 14155:2020, and EU Regulation No. 536/2014. Furthermore, 21 CFR Parts 50, 56, 312, and 812 are US FDA requirements that ensure the protection of human subjects and regulatory compliance for drug and medical device trials.
A clinical QMS operates through a tiered documentation structure comprising a quality manual, quality policy, SOPs, work instructions, and records.
According to the TransCelerate model, a robust clinical QMS is built on foundational aspects, core elements, and management review and assessment. Core QMS elements include processes, resources, roles and responsibilities, partnering, and risk management. Other core elements are issue management, knowledge management, and documentation.
A clinical QMS ensures participant safety and data integrity, while pharmaceutical and medical device QMSs focus on product lifecycle and design-to-market quality, and each QMS follows distinct requirements. Clinical QMS aligns with ICH E6 (R2), ISO 14155, and EU 536/2014. Pharmaceutical QMS aligns with requirements such as ICH Q10 and 21 CFR Parts 210/211. Medical devices are governed by requirements like ISO 13485 and 21 CFR Part 820.
Clinical companies can utilize an electronic Quality Management System (eQMS), which streamlines QMS processes, enhances traceability, and helps ensure compliance with applicable requirements.
SimplerQMS is a fully validated, cloud-based eQMS that streamlines compliance with clinical requirements like ICH E6 (R2), ISO 14155, and EU 536/2014 by centralizing document control and streamlining CAPA, training, audit management, and other key processes.
What Is a Clinical Quality Management System?
A Clinical Quality Management System (CQMS) is a structured framework designed to ensure that clinical research is conducted in accordance with applicable regulatory, scientific, and ethical requirements.
Clinical QMS integrates processes, policies, procedures, and tools that ensure clinical trials are conducted with integrity, regulatory compliance, and a commitment to participant safety.
Clinical QMS provides a basis for consistent quality oversight throughout the clinical trial lifecycle, from early-phase studies to post-marketing surveillance. Key functions include overseeing protocols, managing documentation, facilitating audits, supporting staff training, enhancing risk management, and ensuring regulatory compliance.
Clinical QMS’s scope spans the full range of clinical trial activities and impacts cross-functional areas like project management, data integrity, pharmacovigilance, and regulatory affairs. Clinical QMS supports organizations such as pharmaceutical, biotechnology, medical device companies, healthcare institutions, and contract research organizations (CROs) in managing the quality of clinical operations.
Integrating clinical QMS into operations helps increase efficiency, improve risk mitigation, and ensure patient safety and data reliability. QMS fosters a culture of proactive quality management that reduces errors and ensures trials adhere to requirements such as Good Clinical Practice (GCP), FDA, and European Medicines Agency (EMA).
A quality management system can take various forms based on organizational needs, resources available, or the complexity of an organization. For instance, small CROs may use paper-based QMS format, while larger CRO companies may deploy digital/electronic QMS systems. Quality management software for clinical operations, such as SimplerQMS, streamlines processes, enhances compliance and supports comprehensive, scalable implementation.
What Is Quality in a Clinical Setting?
Quality in a clinical setting or clinical quality means producing reliable, reproducible, scientifically valid data while protecting trial participants. Clinical quality entails clinical operations to maintain quality in design and execution to meet regulatory, ethical, and scientific requirements.
Quality dimensions in a clinical setting include data integrity, protocol compliance, patient safety, regulatory adherence, and operational consistency. These quality dimensions interconnect across protocol development, site management, data collection, monitoring, and reporting, with each function responsible for delivering accurate, timely, and verifiable outputs.
Assuring quality involves implementing quality control (QC) and quality assurance (QA) through independent evaluations at clinical operational steps. These evaluations are supported by tools such as risk-based monitoring, deviation tracking, root cause analysis, and corrective and preventive actions (CAPA). The evaluations are measured using quality metrics such as protocol deviation rates, data query resolution times, audit findings, and inspection outcomes.
Quality is essential in clinical trials because the data collected determines whether treatments are safe and effective. Poor quality can lead to patient harm, regulatory setbacks, loss of trust, and costly delays in sales.
What are the Benefits of Implementing a Clinical Quality Management System?
The benefits of implementing a QMS in a clinical setting are listed below.
- Patient and Consumer Safety: Clinical QMS helps ensure the generation of reliable data that your organization, your partners, regulatory and healthcare authorities, and other stakeholders can use to improve patients’ lives. Clinical QMS helps guarantee clinical trial participants that the generated data will be used solely to advance scientific knowledge. Furthermore, the end-users of the clinical research results are assured that the medications/treatments they receive are based on proper research.
- Regulatory Compliance: A clinical QMS streamlines regulatory compliance by methodically and universally managing quality throughout clinical research. Clinical QMS will range from first-in-human trials to post-marketing studies, hence the assurance of compliance with all the applicable regulatory requirements.
- Quality Assurance: A clinical QMS supportsquality assurance by reducing or eliminating repetitive quality issues. These quality issues include intentional/unintentional non-compliance, insufficient staff training, reduced resources, insufficient supervision by management, lack of protocol clarity, and insufficient quality control in data collection and recording. These quality issues can compromise data integrity and patient safety.
- Holistic Quality Management: Clinical QMS helps organizations systematically and holistically manage quality across the entire range of clinical activities. Clinical QMS further allows organizations to achieve both company objectives and clinical research.
- Proactive Risk Management: Clinical QMS helps organizations proactively deal with issues through predictive analytics, highlighting risk trends before they evolve into issues. The organization can allocate resources efficiently, reduce delays, and maintain audit readiness by incorporating real-time monitoring and early-warning systems.
- Data Integrity: Ensures that Trial Master File (TMF) content is traceable, attributable, and securely maintained.
What Are the Requirements for a CQMS?
The main regulatory requirements for clinical QMS are listed below.
- ICH E6 (R2) Good Clinical Practice: A harmonized guideline for ensuring ethically and scientifically sound clinical trials, safeguarding participant rights and integrity of trial data.
- ISO 14155:2020 Clinical investigation of medical devices for human subjects – Good clinical practice: An international standard defining good clinical practice for medical device investigations, focusing on ethical principles, patient safety, and reliable data.
- Regulation (EU) No 536/2014 on clinical trials on medicinal products for human use: An EU regulation streamlining clinical trial processes, emphasizing participant protection, data transparency, and consistent standards across Member States.
- 21 CFR Part 50 Protection of Human Rights: US FDA regulation that governs the protection of human subjects.
- 21 CFR Part 56 Institutional Review Boards (IRBs): US FDA regulation that governs the operation of IRBs in clinical research involving drugs, biologics, and medical devices.
- 21 CFR Part 312 Investigational New Drug Applications (IND): US FDA requirement that specifies the regulatory requirements for sponsors and investigators seeking FDA approval to conduct clinical trials of investigational new drugs in humans.
- 21 CFR Part 812 Investigational Device Exemptions (IDE): US FDA requirement that governs clinical investigations of unapproved medical devices, permitting their use and distribution under specific FDA regulatory controls.
ICH E6 (R2) Good Clinical Practice
ICH E6 (R2) Good Clinical Practice (GCP) is a harmonized guideline for designing, conducting, recording, and reporting clinical trials involving human subjects. ICH E6 (R2) GCP was developed by the International Council for Harmonization of Technical Requirements for Pharmaceuticals for Human Use (ICH) to ensure high-quality clinical research that protects trial participants and produces credible data.
ICH E6 (R2) is adopted by regulatory authorities in the European Union, the United States, Japan, Canada, and Switzerland, making it a common standard across regions. The guideline promotes global consistency and mutual acceptance of clinical trial results.
The main goal of ICH E6 (R2) is to support quality and oversight in clinical trials by embedding ethical principles, risk-based quality management, and systematic documentation. These principles help ensure that participants’ rights, safety, and well-being are protected throughout the study.
Some key requirements in ICH E6 (R2) include the following.
- Ethical conduct based on the Declaration of Helsinki (Section 2).
- Comprehensive risk-based quality management (Section 5.0).
- Documentation and validation of computerized systems (Section 1.65, 5.5.3).
- Protection of participant rights and safety (Section 3.1.5).
- Defined roles and responsibilities for sponsors, investigators, and CROs (Section 5.2).
- Systematic monitoring strategies (Section 5.18).
- Proper documentation to confirm trial conduct and data quality (Section 8.1).
ICH E6 (R2) is essential to clinical QMS as it strengthens operational integrity, supports reliable trial outcomes, and builds trust among regulators, sponsors, and the public. Enforcement of ICH E6 (R2) is handled by regulatory agencies like the FDA (US), European Medicines Agency (EMA) (EU), and Pharmaceuticals and Medical Devices Agency (PMDA) (Japan).
ISO 14155:2020
ISO 14155:2020 is an international standard that defines Good Clinical Practice (GCP) for designing, conducting, recording, and reporting clinical investigations involving human subjects. ISO 14155:2020 is mainly used to assess medical devices’ safety and clinical performance.
ISO 14155:2020 guides medical device trials to protect participants, ensure scientific integrity, and produce credible evidence. ISO 14155:2020 applies to pre- and post-market studies, excluding in vitro diagnostics.
Several ISO 14155:2020 key requirements are listed below.
- Implementation of clinical quality management (Section 9.1).
- Risk-based monitoring and control strategies (Sections 6.2, 6.7).
- Requirements for informed consent, ethics committee approval, subject privacy, and data confidentiality (Sections 5.6, 5.8, 7.7).
- Detailed planning through a clinical investigation plan (CIP) (Section 6.4, Annex A).
- Documentation of medical device deficiencies and adverse events (Section 7.4).
- Audit readiness and document control (Sections 7.11, 7.8).
The International Organization for Standardization (ISO) developed ISO 14155:2020. National bodies like the FDA (US), EMA (EU), and Health Canada reference this standard for regulatory evaluations.
ISO 14155:2020 is essential for ensuring compliance and quality in clinical investigations of medical devices. It enables the harmonization of global requirements, facilitates market access, and reduces regulatory risk through standardized documentation and procedures.
Regulation (EU) No 536/2014
Regulation (EU) No 536/2014 is a European Union legal framework that governs clinical trials on medicinal products involving human subjects. Regulation (EU) No 536/2014 replaces Directive 2001/20/EC and directly applies to all EU Member States. The European Medicines Agency (EMA) and national competent authorities within each EU Member State enforce the regulation.
Regulation (EU) No 536/2014 aims to streamline and harmonize the processes for clinical trial approvals across the EU. Regulation (EU) No 536/2014 helps protect trial participants and enhances the reliability and robustness of clinical trial data. It introduces uniform rules on ethics committee involvement, informed consent, safety reporting, and transparency through the EU Clinical Trials Information System (CTIS).
Several key Regulation (EU) No 536/2014 chapters are listed below.
- Chapter I General Provisions: Includes scope, definitions, and general principles.
- Chapter II Authorization Procedure for a Clinical Trial: Contains prior authorization, Submission of application, assessment report, decision on the clinical trial, and withdrawal
- Chapter III Authorization Procedure for a Substantial Modification of a Clinical Trial: Specifies the procedures for submitting and approving significant changes to ongoing clinical trials.
- Chapter IV Application Dossier: Lists required documents and content for initial and modified clinical trial applications.
- Chapter V Protection of Subjects and Informed Consent: Outlines ethical standards and legal requirements for obtaining informed consent from trial participants.
- Chapter VI Start, End, Temporary Halt, and Early Termination: Establishes notification rules and timelines for all phases of a clinical trial’s lifecycle.
- Chapter VII Safety Reporting: Describes how adverse events and safety concerns must be reported and assessed during clinical trials.
- Chapter VIII Conduct, Supervision, Training, and Auxiliary Medicinal Products: Regulates trial execution, sponsor oversight, personnel qualifications, and use of supportive medicines.
- Chapter IX Manufacturing and Import: Covers conditions and authorizations for producing and importing investigational and auxiliary products.
- Chapter X Labelling: Defines investigational and auxiliary medicinal product labeling requirements.
- Chapter XI Sponsor and Investigator: Details the responsibilities of sponsors, co-sponsors, legal representatives, and investigators.
- Chapter XII Damage Compensation: Requires member states to have systems ensuring subject compensation for trial-related injuries.
- Chapter XIII Supervision, Inspections, and Controls: Grants member states and the EU authority to inspect and ensure compliance with the regulation.
- Chapter XIV IT Infrastructure: Mandates creating and using an EU portal and database for clinical trial information management.
- Chapter XV Cooperation Between Member States: Establishes mechanisms for coordination and communication between national authorities.
- Chapter XVI Fees: Allows member states to charge fees for clinical trial authorization activities under transparent rules.
- Chapter XVII Implementing and Delegated Acts: Defines the legal basis for future updates and regulatory changes under EU governance.
- Chapter XVIII Miscellaneous Provisions: Addresses topics such as interaction with other EU laws, personal data, and liability.
- Chapter XIX Final Provisions: Details transitional measures, repeal of Directive 2001/20/EC, and implementation timelines.
21 CFR Parts 50 Protection of Human Subjects
21 CFR Part 50 is a US FDA regulation that governs the protection of human subjects. 21 CFR Part 50 regulation ensures that clinical trials protect participant rights, safety, and welfare. It provides the ethical and procedural framework to support quality, integrity, and regulatory compliance in human subject research.
Key requirements include the following.
- Establishes mandatory informed consent procedures (Subpart B).
- Defines exceptions for emergency and minimal risk research (Subpart B Clause 50.22).
- Includes additional safeguards for children in clinical investigations (Subpart D).
- Prohibits waiver of participant legal rights or release from liability (Subpart B Clause 50.20).
21 CFR Part 50 is important for maintaining ethical requirements and ensuring the credibility of clinical research. 21 CFR Part 50 aligns with international ethical guidelines such as the Declaration of Helsinki and supports compliance with ICH E6 (R2).
21 CFR Part 56 Institutional Review Boards (IRBs)
21 CFR Part 56 is a US FDA regulation that governs the operation of IRBs in clinical research involving drugs, biologics, and medical devices. 21 CFR Part 56 ensures participant safety and ethical oversight in clinical trials while providing a framework for quality, integrity, and regulatory compliance.
Several key requirements of 21 CFR Part 56 include the following.
- Requires the establishment and registration of IRBs responsible for ethical review (Subpart B).
- Defines composition, functions, review procedures, and recordkeeping (Subpart B).
- Mandates continuing review of ongoing research (Subpart C).
- Requires that IRBs assess risk-benefit balance and ensure voluntary informed consent (Subpart C).
The US FDA enforces 21 CFR 56 under the Department of Health and Human Services (HHS).
Adherence to 21 CFR 56 ensures that participants’ rights are prioritized and that clinical data can be used to support regulatory approvals.
21 CFR Part 312 (Investigational New Drug Applications)
21 CFR Part 312 outlines the regulatory requirements for sponsors and investigators to obtain US FDA approval to conduct clinical trials involving investigational new drugs in human subjects.
21 CFR Part 312 ensures the protection of trial participants, the scientific validity of collected data, and proper regulatory oversight before investigational drugs can be considered for marketing. 21 CFR Part 312 is foundational in maintaining quality and compliance throughout the clinical development.
Key components for 21 CFR 312 include the following.
- IND requirements (Subpart B Clauses 312.20–23)
- Safety reporting (Subpart B Clause 312.32)
- Annual reporting (Subpart B Clause 312.33)
- Responsibilities of sponsors and investigators (Subpart D Clauses 312.50–70)
- Clinical hold and termination (Subpart C Clauses 312.42, 312.44)
21 CFR Part 312 is crucial for embedding safety, ethical conduct, and data reliability within a clinical QMS. 21 CFR Part 312 provides the legal foundation for all clinical trials involving unapproved drugs in the United States. This requirement aligns with GCP requirements such as ICH E6 (R2).
21 CFR Part 812 (Investigational Device Exemptions)
21 CFR Part 812 regulates clinical investigations of unapproved medical devices, allowing their lawful use and shipment under defined FDA controls.
21 CFR Part 812 ensures investigational medical devices are tested through scientifically sound trials while protecting participant rights, making it essential for clinical quality and compliance.
Several components of 21 CFR Part 812 include the following.
- IDE Application (Subpart B Clauses 812.20–30)
- IRB and FDA Approval (Subpart C Clauses 812.42, 812.62)
- Labeling and Promotion Restrictions (Subpart A Clauses 812.5, §812.7)
- Monitoring and Compliance (Subpart C Clause 812.46)
- Recordkeeping and Reporting (Subpart G Clauses 812.140–150)
- Distinction Between Significant and Non-Significant Risk Devices (Subpart B Clause 812.20)
The FDA’s Center for Devices and Radiological Health (CDRH) enforces 21 CFR Part 812 to regulate trials of unapproved medical devices, ensuring scientific accuracy, ethical conduct, and subject protection. 21 CFR Part 812 supports clinical QMS through documentation, oversight, and alignment with Parts 50 and 56, ensuring valid data and regulatory compliance.
What Is the Documentation Structure of a QMS in Clinical Operations?
A clinical quality management system follows a structured documentation hierarchy consisting of tiered levels of documents designed to support quality, compliance, and operational control across clinical operations.
A tiered documentation system in a clinical QMS ensures consistency, traceability, and regulatory compliance by linking high-level policies to operational procedures and records. This documentation structure creates a clear, auditable framework that supports quality and continuous improvement.
The typical structure of QMS documentation in a clinical QMS is listed below.
- Quality Manual: The quality manual defines the structure, scope, and core processes of the QMS. Although it is not specifically required by ICH E6 (R2), ISO 14155:2020, or Regulation (EU) No 536/2014, many organizations voluntarily adopt it to clearly define their QMS framework and demonstrate their commitment to quality.
- Policies and Objectives: The QMS policies (e.g., quality policies, company policies) and objectives reflect the organization’s commitment to quality, risk management, and regulatory compliance.
- Procedures or Standard Operating Procedures (SOPs): SOPs are high-level instructions detailing how to implement processes.
- Work Instructions (WIs): WIs are task-specific instructions that expand SOPs into detailed steps.
- Records: Training records, monitoring visit reports, deviation logs, and trial master file forms provide documented evidence of performed activities and decisions.
The standard documentation structure in a clinical QMS is shown in the image below.
What is the Clinical Quality Management System Framework?
The clinical QMS framework outlines the foundational and operational elements that uphold quality throughout clinical development. The paper “TransCelerate’s Clinical Quality Management System: From a Vision to a Conceptual Framework” presents a universal model applicable to all clinical research organizations and highlights essential components for a robust clinical QMS.
According to TransCelerate’s Clinical Quality Management System: From a Vision to a Conceptual Framework, the clinical QMS framework includes the items listed below.
- Foundational Aspects: Define the environment, leadership, governance, and culture needed to support a clinical QMS.
- Core Elements of a Clinical QMS Framework: Address quality across all levels and functions of clinical development.
- Management Review and Assessment of the Clinical QMS: Evaluate QMS effectiveness, identify improvement areas, and adapt based on internal and external changes.
The clinical QMS framework components are summarized and shown in the image below.
Foundational Aspects
Foundational aspects outline the leadership, governance structure, organizational culture, and supportive environment essential for implementing and maintaining a clinical QMS.
The foundational aspects of clinical QMS include the following.
- Understanding of the Concept: Recognize the internal and external factors, such as regulatory requirements, organizational structure, and past experiences, that influence how the clinical QMS operates.
- Leadership Commitment to Quality: Senior management defines and communicates clear and consistent expectations that set the tone for a quality-focused culture.
- Organizational Commitment to Quality: The entire organization shares responsibility for quality, where all employees are empowered to speak up, raise concerns, and take proactive action.
- Continuous Improvement: The clinical QMS continually evolves to enhance effectiveness and efficiency.
Understanding of the Concept
Understanding a clinical QMS involves evaluating internal and external factors shaping how the system is designed, implemented, and maintained. This essential aspect ensures the QMS is tailored to the organization’s unique environment, aligned with regulatory expectations, and fully fit for purpose.
External factors include global regulatory requirements, laws, market conditions, and stakeholder expectations. Internal factors involve the organization’s structure, culture, strategic goals, and historical performance. External and internal insights enable the customization of QMS processes to support compliance and performance better.
This foundational understanding of the concept strengthens key QMS elements such as risk management, issue management, and knowledge management. While not always explicitly mandated, frameworks like ICH E6 (R2), ISO 14155:2020, and Regulation (EU) No. 536/2014 depend on this understanding to ensure the QMS remains practical, adaptable, and compliant in real-world clinical settings.
Leadership Commitment to Quality
Leadership commitment in a clinical QMS involves the active engagement of senior management in fostering a culture of quality across all levels of the organization.
Leadership commitment ensures quality is embedded across all roles and processes, not confined to a single department. Leaders set the tone by defining policies and objectives and making decisions aligned with regulatory requirements.
Strong leadership enables key QMS components like risk management, issue management, and training by ensuring access to resources, qualified personnel, and robust governance structures. Regulatory frameworks such as ICH E6 (R2), ISO 14155:2020, and Regulation (EU) No. 536/2014 explicitly expect visible and documented leadership engagement in QMS.
Organizational Commitment to Quality
Organizational commitment to quality in a clinical QMS means all employees are responsible for embedding quality into their daily work and decision-making.
The organizational commitment to quality promotes a culture where staff are empowered to raise concerns, drive improvements, and uphold quality through open communication and cross-functional collaboration.
Such commitment strengthens internal controls, helps maintain regulatory compliance, and enhances core QMS components like risk and deviation management. ICH E6 (R2), ISO 14155:2020, and Regulation (EU) No. 536/2014 emphasize this principle, reinforcing the importance of a proactive, organization-wide quality culture that supports clinical trial integrity and success.
Continuous Improvement
Continuous improvement in a clinical QMS is the structured, ongoing process of identifying and applying enhancements to strengthen quality, compliance, and performance across clinical operations.
Continuous improvement helps ensure the QMS adapts to internal changes, stakeholder needs, and regulatory expectations. Continuous improvement is achieved through regular assessments, internal audits, root cause analysis, employee feedback, and trend reviews of quality data. These tools help uncover inefficiencies, risks, or compliance issues and drive targeted improvements across the system and within specific processes.
Key QMS areas such as issue management, risk management, and knowledge management are some of the processes that continuous improvement supports. Continuous improvement is aligned with the expectations of requirements like ICH E6 (R2), ISO 14155:2020, and Regulation (EU) No. 536/2014.
Core Elements of a Clinical QMS Framework
Core elements of a clinical QMS framework are the operational areas that collectively ensure a clinical QMS is effective, efficient, and fit for purpose.
The different core elements of a clinical QMS framework are listed below.
- Processes: Establish clear workflows and procedural documents (e.g., SOPs) to guide consistent and compliant execution of clinical activities.
- Resources, Roles, and Responsibilities: Assign qualified personnel, tools, and infrastructure with clearly defined responsibilities and accountability.
- Partnering: Integrate external partners such as contract research organizations (CROs) and vendors to ensure alignment with requirements and expectations.
- Risk Management: Identify, assess, and mitigate risks impacting clinical quality, compliance, or patient safety.
- Issue Management: Detect, investigate, and resolve issues, including conducting root cause analysis and implementing corrective and preventive actions (CAPA).
- Knowledge Management: Capture, share, and use organizational knowledge to support decision-making and reduce the likelihood of repeated errors.
- Documentation Supporting Achievement of Quality: Control and organize documents and records (e.g., policies, SOPs, forms, and reports) that provide evidence of compliance with quality and regulatory requirements.
Processes
Processes are structured sequences of actions that guide how clinical tasks should be performed, ensuring consistency, compliance, traceability, and accountability.
Processes typically follow a lifecycle of initiation, review, approval, execution, and closure. Documents such as policies, SOPs, and work instructions ensure clarity and standardization, reflecting leadership’s commitment to quality.
Defined processes promote quality, regulatory compliance, and operational efficiency. Processes clarify roles, support training, and reduce variability in execution.
Processes are pivotal to the successful functioning of a clinical QMS and are closely integrated with other key elements, such as risk management, issue management, and documentation control. Processes provide the structure through which regulatory requirements, such as GCP, are operationalized and monitored.
Resources, Roles, and Responsibilities
Resources refer to the personnel, systems, and tools needed to implement QMS. Roles and responsibilities define who is accountable for specific tasks across the organization.
Resources, roles, and responsibilities may begin with identifying required skillsets and resources, followed by assigning roles, providing training, and ensuring qualifications align with task requirements. Responsibilities are regularly reviewed and updated to match operational needs and expectations.
Clear roles and adequate resources ensure consistent execution, regulatory compliance, and operational efficiency across clinical activities. Resources, roles, and responsibilities support key QMS functions like training, document control, and audits.
Partnering
Partnering refers to structured collaboration with external parties such as vendors, CROs, and suppliers, in which all involved parties share responsibility for maintaining quality and compliance.
The partnering process typically includes vendor selection, qualification, agreement on roles and expectations, performance monitoring, and periodic re-evaluation or requalification.
Effective partnering ensures that external contributors meet defined requirements and support operational efficiency. Partnering ties into QMS elements like supplier management, documentation control, and audit readiness.
Risk Management
Risk management is the structured process of identifying, assessing, controlling, and monitoring potential uncertainties that could impact clinical trial quality, patient safety, or regulatory compliance. This proactive approach aims to detect risks early before they interfere with operational goals or stakeholder expectations.
The risk management lifecycle includes several key stages such as identifying potential risks, evaluating their likelihood and severity, and documenting risk information for traceability. Additionally, control measures should be implemented to mitigate the impact and continuously monitor and review risks throughout the lifecycle of the clinical process or product.
By integrating risk management into clinical activities, organizations foster a quality, accountability, and readiness culture. Furthermore, risk management connects with other QMS components such as issue management, design control, and supplier management.
Issue Management
Issue management refers to identifying, investigating, and resolving issues (e.g., deviation, nonconformance, complaint, audit finding) that could affect patient safety, data integrity, or regulatory compliance.
Issue management is vital for addressing deviations, errors, or non-conformities that arise during clinical operations. The issue management lifecycle typically includes detecting the issue, assessing its impact, assigning responsibility and an initial plan, and investigating, often involving root cause analysis. This is followed by implementing CAPAs, assessing their effectiveness, and formally closing the issue with continued monitoring to ensure effective resolution.
Effective issue management enables a timely and organized response to issues, reduces the likelihood of recurrence, and strengthens overall compliance and operational efficiency. Issue management supports clinical integrity and builds trust with regulators, partners, and stakeholders. This function is closely linked to other QMS elements such as CAPA, audits, risk management, and documentation control.
Knowledge Management
Knowledge management refers to the process of capturing, organizing, and distributing critical information to ensure that employees have timely access to the knowledge needed to perform their roles effectively and in compliance with regulatory requirements.
The knowledge management lifecycle typically involves identifying essential knowledge areas and creating and reviewing relevant content such as standard operating procedures (SOPs). Training materials, if available, are trained and distributed, evaluation of staff understanding, and training records are maintained for traceability and updates.
Effective knowledge management reduces errors, prevents gaps in execution, and strengthens regulatory compliance by keeping personnel informed and capable. A failure in knowledge sharing, such as assigning untrained personnel to critical tasks, can compromise patient safety, data integrity, and overall trial reliability.
Knowledge management directly supports QMS components such as training, document control, and audit readiness.
Documentation Supporting Achievement of Quality
Documentation supporting the achievement of quality includes policies, SOPs, work instructions, forms, and records like reports. Documents guide daily operations and ensure consistency in clinical processes, and records provide evidence of compliance.
The documentation lifecycle includes document creation, review, approval, distribution, execution, and retirement. The documentation lifecycle determines how long and where documents must be stored to meet regulatory and operational needs.
Accurate and accessible documentation ensures that clinical activities are traceable, consistent, and aligned with quality and regulatory requirements. Documentation is central to all QMS elements, enabling effective training, risk management, and issue management.
Management Review and Assessment of the Clinical QMS
Management review and assessment of the clinical QMS is a structured activity by senior management to evaluate its effectiveness, suitability, and efficiency.
Management review and assessment help ensure the clinical QMS meets quality objectives and supports compliance and operational performance.
The two main aspects of management review and assessment are listed below.
- Assessment of the Clinical QMS: Involves evaluating each core QMS element individually and in aggregate.
- Management Review: Requires senior leadership with decision-making authority to review QMS performance regularly.
Assessment of the Clinical QMS
Assessment of the clinical QMS involves evaluating both individual and integrated QMS elements to ensure they function effectively and align with quality objectives and regulatory expectations.
The assessment process typically includes a baseline evaluation of the current state, followed by gap identification, plan improvement, and implementation of improvement actions. These assessments feed and recommend actions for strategic oversight in the management review.
Regular assessments are essential for identifying gaps, measuring system performance, and driving continuous improvement. QMS assessments support and inform elements like risk management, CAPA, training, and audits.
Management Review
The management review refers to the formal, periodic evaluation of the clinical QMS by senior leadership to ensure it remains effective, aligned with quality objectives, and supports continuous improvement.
The management review process typically involves planning, data collection (e.g., KPIs, audit results, CAPAs), review meetings, documentation of outcomes, and implementation of follow-up actions. This cycle is repeated at defined intervals to maintain oversight.
Management review is essential for verifying that the clinical QMS meets company objectives and ensures product and process quality. Additionally, management review drives informed decision-making, resource allocation, and system improvements.
Management review connects with all core QMS elements such as risk management, audit management, training management, and document control.
How Does a Clinical QMS Differ From a Pharmaceutical and Medical Device QMS?
A clinical QMS differs from a pharmaceutical and medical device QMS in its area of focus, regulatory frameworks, and output.
A clinical QMS focuses on managing quality throughout clinical trials, supporting compliance and oversight during clinical research and development. A pharmaceutical QMS manages quality throughout the product lifecycle, from drug formulation to manufacturing, packaging, and distribution. A medical device QMS, on the other hand, focuses on the design, manufacturing, labeling, traceability, and post-market surveillance of medical devices.
A clinical QMS’s primary focus is on subject safety and data reliability, while pharmaceutical QMS and medical device QMS emphasize product quality, safety, and manufacturing consistency.
Moreover, the regulatory landscape varies. Clinical QMS is guided by ICH E6 (R2), ISO 14155, and EU Regulation No. 536/2014 frameworks. Pharmaceutical QMS aligns with requirements like ICH Q10 and US FDA 21 CFR 210/211. Medical device QMS follows requirements such as ISO 13485 and FDA 21 CFR Part 820.
While a clinical QMS focuses on demonstrating credibility, a clinical QMS can be integrated with a pharmaceutical or medical device QMS to support the management of the TMF and ensure alignment across regulatory requirements.
What Is the Role of QMS Software in Clinical Operations?
QMS software plays an important role in clinical operations by enabling efficient, traceable, and compliant execution of quality-related processes, including TMF creation and maintenance throughout the entire clinical trial lifecycle. QMS software replaces manual, paper-based systems with centralized digital platforms that enhance accuracy, automate workflows, and support regulatory compliance.
QMS software automates core quality processes such as document control, training management, change control, deviation tracking, audit readiness, CAPA, supplier qualification, risk management, and more.
Additionally, QMS software offers real-time access, full traceability, electronic signatures, automated task assignments, and streamlining activities from pre-trial planning through study closure.
SimplerQMS is a fully validated eQMS solution designed for life science companies. SimplerQMS meets FDA 21 CFR Part 11 and EU Annex 11 requirements and is validated to GAMP5 requirements. Furthermore, SimplerQMS supports compliance with ICH E6 (R2), ISO 14155, and Regulation (EU) No. 536/2014.
SimplerQMS can centralize clinical documentation such as protocols, informed consent forms, investigator brochures, and trial master files. SimplerQMS can further manage training, suppliers, audits, and more.